WO2015079213A1 - Hif inhibitors - Google Patents

Hif inhibitors Download PDF

Info

Publication number
WO2015079213A1
WO2015079213A1 PCT/GB2014/053475 GB2014053475W WO2015079213A1 WO 2015079213 A1 WO2015079213 A1 WO 2015079213A1 GB 2014053475 W GB2014053475 W GB 2014053475W WO 2015079213 A1 WO2015079213 A1 WO 2015079213A1
Authority
WO
WIPO (PCT)
Prior art keywords
hif
compound
pharmaceutically acceptable
solvate
derivative
Prior art date
Application number
PCT/GB2014/053475
Other languages
French (fr)
Inventor
Margaret ASHCROFT
Keith Jones
Original Assignee
Ucl Business Plc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ucl Business Plc filed Critical Ucl Business Plc
Priority to US15/039,358 priority Critical patent/US20170157112A1/en
Priority to EP14803226.1A priority patent/EP3074013A1/en
Priority to JP2016534721A priority patent/JP2016538304A/en
Publication of WO2015079213A1 publication Critical patent/WO2015079213A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4745Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having nitrogen as a ring hetero atom, e.g. phenantrolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2121/00Preparations for use in therapy

Definitions

  • the invention relates to hypoxia-inducible factors (HIF), and particularly, although not exclusively, to the inhibition of HIF activity.
  • HIF hypoxia-inducible factors
  • the invention extends to inhibitors of HIF activity, and their use in the prevention or inhibition of diseases characterised by abnormal HIF activity or levels, such as tumour progression, and the treatment of cancer.
  • the invention encompasses pharmaceutical compositions and methods of treating diseases characterised by elevated HIF activity, such as cancer.
  • hypoxia-inducible factor (HIF) transcriptional complex is involved in tumour progression by up-regulating key genes involved in metabolic adaptation, glycolysis (glucose transporters, GLUTi and glycolytic enzymes), proliferation (insulin-like growth factors l and 2) and angiogenesis (VEGF, erythropoietin).
  • HIF is a dimeric transcription factor comprising a regulatory a subunit and constitutively expressed ⁇ subunit.
  • HIF-a availability is controlled at the level of protein stability and synthesis by changes in oxygen concentration and growth factors, respectively.
  • Over- expression of HIF-a occurs in most human cancers due to changes in micro- environmental stimuli (e.g. hypoxia, growth factors) and genetic abnormalities that lead to loss of tumour suppressor function (e.g. p53, PTEN, VHL) or oncogenic activation (e.g. Ras, Myc, Src).
  • the inventors have developed a cell-based reporter screen (known as "U2OS-HRE- luc") that was used to identify novel small molecule inhibitors of HIF activity. Using this assay, they have now found that one of their hit compounds (which is referred to herein as the compound represented by formula I or simply "formula I” or "HIF-
  • Inhibi inhibits both HIF activity and HIF-a expression in response to hypoxia and growth factors in several cancer cell lines. As such, they are the first group to have demonstrated a therapeutic use for the lead compound, which can be used in the treatment or prevention of cancer.
  • the inventors propose that compound of formula (I) also has use in other settings where blockade of HIF is therapeutically beneficial (e.g. in the hepatitis C viral (HCV) infection life cycle and hepatoma cell migration).
  • HCV hepatitis C viral
  • HIF hypoxia-inducible factor
  • a method of treating, preventing or ameliorating a disease characterised by abnormal levels of hypoxia-inducible factor (HIF) activity preferably cancer
  • the method comprising administering, to a subject in need of such treatment, a therapeutically effective amount of a compound of formula (I), or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof.
  • HIF hypoxia-inducible factor
  • the inventors have shown that the compound of formula (I) not only effectively inhibits HIF activity, but also HIF-a expression in response to hypoxia and growth factors in several cancer cell lines.
  • compound (I) inhibits the growth of a panel of tumour cell lines at submicromolar concentrations. Evaluation of compound (I) showed that it has favourable pharmacokinetic properties in vivo and mice could tolerate a maximum dose of up to loomg/kg daily dosing by intraperitoneal (IP) injection.
  • IP intraperitoneal
  • compound (I) is structurally similar to emetine, a known protein synthesis inhibitor.
  • emetine a known protein synthesis inhibitor.
  • the inventors have found that compound (I) is at least 100-fold less toxic than emetine on tumour cells.
  • previous studies have shown that emetine targets the 40S ribosome at the level of the ribosomal protein S14.
  • the compound of formula (I), or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof inhibits the hypoxia-inducible factor (HIF) transcriptional complex, i.e. it is a HIF pathway inhibitor.
  • HIF hypoxia-inducible factor
  • the compound of formula (I), or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof reduces or blocks expression of hypoxia-inducible factor-i alpha (HIF- ⁇ ).
  • HIF- ⁇ hypoxia-inducible factor-i alpha
  • the compound of formula (I), or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof reduces or blocks expression of vascular endothelial growth factor (VEGF).
  • VEGF vascular endothelial growth factor
  • the compound's potency in blocking VEGF induction in hypoxia directly correlates with its IC 50 for inhibiting HIF activity, i.e. ⁇ .25- ⁇ .5 ⁇ .
  • the compound of formula (I), or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof reduces or blocks eIF-2a phosphorylation.
  • the compound's potency in blocking eIF-2a phosphorylation directly correlates with its IC 50 for inhibiting HIF activity, i.e. ⁇ .25- ⁇ .5 ⁇ .
  • compound of formula (I), or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof can be used to treat any disease resulting from abnormal levels of HIF or HIF activity.
  • abnormal HIF levels may be decreased with respect to those in a healthy individual.
  • the disease is characterised by elevated HIF activity with respect to a healthy individual.
  • HIF is constitutively upregulated and HIF-a (HIF-ia or HIF-2a) protein is overexpressed.
  • HIF-a HIF-ia or HIF-2a
  • HCV hepatitis C viral infection life cycle is known to result in elevated HIF activity, and so hepatitis C can be treated using the compound of formula (I), or a functional analogue, pharmaceutically acceptable salt or solvate thereof.
  • pharmaceutically acceptable salt or solvate thereof can be used to treat any tumour or cancer-based disease where HIF is constitutively upregulated and HIF-a (HIF-ia or HIF-2a) protein is overexpressed.
  • the cancer may be a solid tumour or solid cancer.
  • compound of formula (I), or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof, is used to treat prostate cancer. Hepatoma cell migration may also be treated.
  • a functional analogue can be defined as being any compound which exhibits at least 8o% HIF inhibition compared to compound (I) using the U20S-HRE-luc cell-based assay without affecting cell viability, i.e. the analogue is not toxic.
  • Toxicity can be defined as being more than 20% cell death within 24 hours, and so functional analogues should not cause more than 20% death.
  • analogues of compound (I) are shown in Figures 7-12.
  • the chemical structure of compound (I) can be broken down into three subunits as shown by the double lines in the centre of Figure 8.
  • Arrows ⁇ and 3 in Figure 8 indicate that there are up to 6-iiindependent chemical groups in combination with up to three separate cores resulting in a variety of functional analogues.
  • preferred analogues of compound (I) are shown in Figure 8.
  • Compound (I), for use, in the invention, may be chiral.
  • the compound (I) may include any diastereomer and enantiomer of the formula represented by (I).
  • Diastereomers or enantiomers of (I) are believed to display potent HIF inhibitory activity, and such activities may be determined by use of appropriate in vitro and in vivo assays, which will be known to the skilled technician.
  • Compounds defined by formula (I) can therefore include analogues as racemates.
  • the compounds of formula (I) can be pairs of diastereoisomers, or individual
  • enantiomers including the threo- and eryi ro-pair of diastereoisomers and the individual threo and erythro enantiomers.
  • the compound (I) is the S, R enantiomer, i.e. (S)-2-(((R)-6,7-dimethoxy- i,2,3,4-tetrahydroisoquinolin-i-yl)methyl)-3-ethyl-i,6,7,iib-tetrahydro-4H- pyrido[2,i-a]isoquinoline.
  • compounds for use in the invention may also include pharmaceutically active salts, e.g. the hydrochloride.
  • the compound of formula (I) is a surprisingly effective HIF pathway inhibitor.
  • hypoxia-inducible factor (HIF) pathway inhibitor comprising a compound of formula (I), or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof.
  • a compound of formula (I), or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof for use as hypoxia-inducible factor (HIF) pathway inhibitor.
  • the compound of formula (I), or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof according to the invention maybe used in a medicament which may be used in a monotherapy (i.e. use of compound (I) alone), for treating, ameliorating, or preventing a disease characterised by abnormal levels of hypoxia-inducible factor (HIF) activity, preferably cancer.
  • the compound of formula (I), or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof according to the invention may be used as an adjunct to, or in combination with, known therapies for treating, ameliorating, or preventing cancer.
  • compositions having a number of different forms depending, in particular, on the manner in which the composition is to be used.
  • the composition maybe in the form of a powder, tablet, capsule, liquid, ointment, cream, gel, hydrogel, aerosol, spray, micellar solution, transdermal patch, liposome suspension or any other suitable form that may be administered to a person or animal in need of treatment.
  • vehicle of medicaments according to the invention should be one which is well -tolerated by the subject to whom it is given.
  • Medicaments comprising the compound of formula (I), or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof according to the invention may be used in a number of ways. For instance, oral
  • compositions comprising the compounds of the invention may be administered by inhalation (e.g. intranasally).
  • Compositions may also be formulated for topical use. For instance, creams or ointments may be applied to the skin.
  • Compounds according to the invention may also be incorporated within a slow- or delayed-release device. Such devices may, for example, be inserted on or under the skin, and the medicament may be released over weeks or even months.
  • the device maybe located at least adjacent the treatment site. Such devices may be particularly advantageous when long-term treatment with compounds used according to the invention is required and which would normally require frequent administration (e.g. at least daily injection).
  • compounds and compositions according to the invention may be administered to a subject by injection into the blood stream or directly into a site requiring treatment. Injections maybe intravenous (bolus or infusion) or subcutaneous (bolus or infusion), or intradermal (bolus or infusion).
  • the amount of the compound that is required is determined by its biological activity and bioavailability, which in turn depends on the mode of administration, the physiochemical properties of the compound, and whether it is being used as a monotherapy, or in a combined therapy.
  • the frequency of administration will also be influenced by the half-life of the compound within the subject being treated.
  • Optimal dosages to be administered may be determined by those skilled in the art, and will vary with the particular compound in use, the strength of the pharmaceutical composition, the mode of administration, and the advancement of the cancer. Additional factors depending on the particular subject being treated will result in a need to adjust dosages, including subject age, weight, gender, diet, and time of administration.
  • a daily dose of between o.o ⁇ g/kg and 500mg/kg of body weight, or between o.img/kg and 200mg/kg body weight of the compound according to the invention may be used for treating, ameliorating, or preventing cancer depending upon which compound or analogue is used.
  • the compound may be administered before, during or after onset of the cancer to be treated.
  • Daily doses may be given as a single administration (e.g. a single daily injection).
  • the cancer may require administration twice or more times during a day.
  • compound (I) may be administered as two (or more depending upon the severity of the cancer being treated) daily doses of between 25mg and 7000 mg (i.e. assuming a body weight of 70 kg).
  • a patient receiving treatment may take a first dose upon waking and then a second dose in the evening (if on a two dose regime) or at 3- or 4-hourly intervals thereafter.
  • a slow release device may be used to provide optimal doses of the compounds according to the invention to a patient without the need to administer repeated doses.
  • a pharmaceutical composition comprising a compound of formula (I), or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof, and a
  • the pharmaceutical composition can be used in the therapeutic amelioration, prevention or treatment in a subject of a disease characterised by abnormal levels of hypoxia-inducible factor (HIF) activity, preferably cancer.
  • the composition is preferably an anti-cancer pharmaceutical composition.
  • the compound (I) is (S)-2-(((R)-6,7-dimethoxy-i,2,3,4- tetrahydroisoquinolin-i-yl)methyl)-3-ethyl-i,6,7,iib-tetrahydro-4H-pyrido[2,i- a]isoquinoline.
  • the invention also provides in a seventh aspect, a process for making the
  • composition according to the sixth aspect comprising contacting a therapeutically effective amount of a compound of formula (I), or a functional analogue, pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable vehicle.
  • a "subject” may be a vertebrate, mammal, or domestic animal.
  • compounds, compositions and medicaments according to the invention maybe used to treat any mammal, for example livestock (e.g. a horse), pets, or may be used in other veterinary applications. Most preferably, however, the subject is a human being.
  • a “therapeutically effective amount” of compound is any amount which, when administered to a subject, is the amount of drug that is needed to treat the target disease, or produce the desired effect, i.e. inhibits HIF activity.
  • the therapeutically effective amount of compound used maybe from about o.oi mg to about 8oo mg, and preferably from about o.oi mg to about 500 mg.
  • a "pharmaceutically acceptable vehicle” as referred to herein is any known compound or combination of known compounds that are known to those skilled in the art to be useful in formulating pharmaceutical compositions.
  • the pharmaceutically acceptable vehicle may be a solid, and the composition may be in the form of a powder or tablet.
  • the composition may be in the form of a powder or tablet.
  • liquid pharmaceutical vehicle maybe a liquid, and the pharmaceutical composition is in the form of a solution.
  • Liquid pharmaceutical compositions which are sterile solutions or suspensions, can be utilized by, for example, intramuscular,
  • intrathecal, epidural, intraperitoneal, intravenous and particularly subcutaneous injection intrathecal, epidural, intraperitoneal, intravenous and particularly subcutaneous injection.
  • Figure l shows that compound I of the invention (referred to herein as "HIF- Inhibi”) blocks HIF activity and HIF-a protein induction in hypoxia in a dose- dependent manner without affecting HIF- ⁇ or key cellular signaling proteins, ERKi/2 and AKt/PKB.
  • Figure lA Graph shows HIF (HRE-luciferase) activity measured as relative light units (RLU) in U20S-HRE-luc cells in response to HIF- Inhibi treatment over a dose range as indicated in normoxia or hypoxia for 16 hours. U20S-HRE-luc described in A, were harvested for western blot analysis.
  • HIF HRE-luciferase activity measured as relative light units (RLU) in U20S-HRE-luc cells in response to HIF- Inhibi treatment over a dose range as indicated in normoxia or hypoxia for 16 hours.
  • RLU relative light units
  • Figure lB Western blots show the effects of HIF-Inhibi on HIF- ⁇ protein in normoxia or hypoxia. Actin was used as a load control.
  • Figure lC Western blots show HIF-ia, phosphorylated ERK1/2 (ERK1/2-P), and AKT/PKB proteins in the absence (-) and presence of ⁇ HIF-Inhibi in normoxia (norm) or hypoxia (hyp). Actin was used as a load control.
  • Figure lD Western blots show the effects of HIF-Inhibi (luM) on HIF-2a protein levels. UT (untreated), and DMSO treated (-) controls are indicated;
  • Figure 2 shows that HIF-Inhibi blocks the induction of HIF targets (GLUTi and VEGF) and tumour cell migration in hypoxia.
  • Figure 2A Graph shows vascular endothelial growth factor (VEGF) protein expression measured by ELISA in U2OS- HRE-luc cells in response to HIF-Inhibi treatment over a dose range as indicated in normoxia or hypoxia for 16 hours
  • Figure 2B U20S-HRE-luc described in A, were harvested for western blot analysis. Western blots show the effects of HIF-Inhibi on glucose transporteri (GLUTi) protein induction in normoxia or hypoxia. Actin was used as a load control.
  • VEGF vascular endothelial growth factor
  • Figure 2C Graph shows tumour cell migration (number (no) of migrated cells/field of view) in the absence (-) and presence of 0.5 or 2.5 ⁇ HIF- Inibi in normoxia (norm) or hypoxia (hyp) for 16 hours;
  • Figure 3 shows that HIF-Inhibi targets the protein translation machinery.
  • Figure 3A Western blots show the effects of HIF-Inhibi treatment over a dose range on HIF- ⁇ and phosphoryated eIF-20: (eIF-2a-P) proteins in U20S-HRE-luc cells in normoxia or hypoxia for 16 hours. Actin was used as a load control.
  • Figure 3B shows tumour cell migration (number (no) of migrated cells/field of view) in the absence (-) and presence of 0.5 or 2.5 ⁇ HIF- Inibi in normoxia (norm) or hypoxia (hyp) for 16 hours;
  • Figure 3 shows that HIF-Inhibi targets the protein translation machinery.
  • Figure 3A Western blots show the
  • Figure 5 shows that HIF-Inhibi blocks HIF-a, VEGF, tumour growth and metastasis (local and distant) in a human PC3 orthotopic mouse xenograft model.
  • Figure 5A Western blots shows the effects of control (solv.con) or HIF-Inhibi treatment on the levels of HIF- ⁇ and HIF- ⁇ proteins PC3 tumour xenografts excised as indicated at day 16 after 75mg/kg daily dosing by intraperitoneal injection.
  • Figure 5B Graph shows VEGF protein levels (pg/ml) from pooled tumour xenographs described in A.
  • Figure 5C-F Graphs show body weight in tumour bearing mice described in A (C), primary tumour weight in grams (g) at day 16 (D), and the weight (g) of local (E) and distant (F) lymph node metastasis.
  • Figure 5G Graph shows levels of HIF-Inhibi ( ⁇ ) in plasma and pooled tumours described in A, measured by LCMS analyses;
  • Figure 6 shows the chemical structure of HIF-Inhibi according to the invention;
  • Figure 7 shows the effects of a series of HIF-Inhibi analogues on HIF activity in U20S-HRE-luc cells.
  • Figure 7A Structures and molecular weights (MW) are shown for a series of chemical analogues (labelled 4-15) of HIF-Inhibi.
  • Figure 7B Graph shows the effects of DMSO control (1), HIF-Inhibi at ⁇ (2), Emetine at ⁇ . ⁇ (3) and analogues (4-15 at ⁇ ) on HIF activity (relative light units) in the U2OS- HRE-luc cell-based assay in hypoxia (1% 0 2 , for 16 hours);
  • Figure 8 shows the structures of a panel of functional analogues of HIF-Inhibi that include a variety of different chemical groups as indicated at positions 1, 2 and 3 within the active phamacophore;
  • Figure 9 shows the reaction scheme for synthesising chemical enantiomers and analogues of HIF-Inhibi;
  • Figure 10 shows the reaction scheme for synthesising 3-dimethylaminomethyl- pentan-2-one methiodide
  • Figure 11 shows the purified structures of five of the chemical enantiomers and analogues of HIF-Inhibi which were obtained by using the reaction scheme shown in Figure 9; and Figure 12 shows the effects of the chemical enantiomers and analogues shown in Figure 11 on HIF activity.
  • Figure 12A is a graph showing the percentage inhibition of luciferase activity in U2OS-HRE cells treated with the compounds shown in Figure 11. The compounds were dosed at ⁇ and incubated in 1% 02 for 16 hours.
  • Figure 12B shows western blot analysis of U2OS-HRE cells treated with compounds indicated including HIF-Inhibi (HIF-Inh) as in Figure lC to show inhibitory effects on HIF- ⁇ , phosphorylated and total eIF2a protein levels. Tubulin was used as a loading control. All data shown has been either averaged or is representative of 3 independent experiments. Examples
  • Example l - Compound I of the invention blocks HIF-ia protein induction in hypoxia U20S-HRE-luc cells were exposed to normoxia or hypoxia (1% 0 2 ) for 16 hours in the presence of DMSO (control) or HIF-Inhibi over a concentration range (0.1- ⁇ ). Cells were harvested and assessed for HRE-luciferase activity as a measure of HIF activity, and for western blot analysis.
  • compound I of the invention blocks HIF activity (HRE- luciferase activity measured as relative light units, RLU) in a dose dependent manner (Figure lA).
  • HIF activity HRE- luciferase activity measured as relative light units, RLU
  • Figure lA This dose-dependent inhibitory effect on HIF activity was found to directly correlate with blockade of HIF-a protein induction in hypoxia ( Figure lB).
  • Figure iC key cellular signaling proteins
  • Figure iC key cellular signaling proteins
  • Example 2 Compound I blocks HIF targets (GLUTi and VEGF) and tumour cell migration in hypoxia
  • U2OS-HRE-IUC cells were exposed to normoxia or hypoxia (1% 0 2 ) for 16 hours in the presence of DMSO (control) or HIF-Inhibi over a concentration range (0.1- ⁇ ).
  • Cells were harvested and assessed for VEGF and GLUTi protein levels using a quantitative ELISA or by western blot analysis respectively.
  • tumour cells were exposed to 0.5 or 2.5 ⁇ HIF-Inhibi in hypoxia, and tumour cell migration was measured using a 2-dimensional filter-based migration assay.
  • Figure 4 shows that the concentrations of compound I between left and right flank subcutaneous tumours were comparable. Tumour concentrations ranged between ⁇ .9-35 ⁇ . Plasma concentrations ranged between 0.07 and 0.3 ⁇ .
  • Example 5 Compound I blocks HIF-q. VEGF. tumour growth and metastasis (local and distant) in a human PCsLNf; orthotopic mouse xenograft model
  • PC3LN5 (10 5 cells) were implanted intraprostatically into mice (Nu) and tumours were allowed to develop for 12 days. Mice received HIF-Inhibi (75mg.kg _1 ) by IP injection daily for 2.5 weeks. Plasma and tumour samples were taken 24h after the last dose and analysed by LCMS. Tumours were excised and homogenised, and assessed for PD endpoints HIF- ⁇ and VEGF proteins. Local and distant lymph node metastases were also evaluated.
  • Figure 5 shows that compound I blocks HIF- ⁇ and VEGF protein in PC3LN5 orthotopic tumours in vivo.
  • Mouse body weight was not significantly affected over 16 days of daily dosing with HIF-Inhibi, indicating minimal toxicity.
  • HIF-Inhibi significantly blocked tumour growth and metastasis (local and distant) in the PC3LN5 orthotopic xenograft model.
  • HIF-Inhibi showed a good PK profile in tumours, indicating good bioavailability to the tumour.
  • a series of analogues (labelled 4-15) of compound I were synthesised, and their structures are shown in Figure 7.
  • U20S-HRE-luc cells were exposed to hypoxia (1% 0 2 ) for 16 hours in the presence of DMSO (control), HIF-Inhibi ( ⁇ ), emetine (0.017UM) as positive control, and then each of the analogues (4)-(i5) as shown in Figure 7.
  • Cells were harvested and luciferase activity was measured in cell lysates using a standard luminometer. Data was represented as relative light units (RLU) for each condition.
  • RLU relative light units
  • a flask was charged with diethyl phthalate (6.9 g, 31 mmol), sodium ethoxide solution (50.3 g, 155 mmol, 21% wt in ethanol), and ethanol (90 mL) and cooled to -5 °C under an atmosphere of nitrogen.
  • Triethyl phosphonoacetate (10.9 g, 49 mmol) was added dropwise maintaining a temperature ⁇ 5 °C.
  • the solution was allowed to warm to 10 °C and stirred for 1 hr before being cooled to o °C.
  • Stage 1 (8.9 g, 39 mmol) was added in one portion and the mixture stirred for 3 hrs at RT followed by 2 hours at reflux.
  • the ethanol was removed in vacuo and the residue partitioned between toluene (400 mL) and water (400 mL). The phases were separated and the aqueous extracted with a further portion of toluene (50 mL). The combined organics were extracted into lM HC1 (500 mL) which was then basified with NaOH and twice extracted into diethyl ether (2 x 400 mL). The organics were dried over MgS04, filtered and concentrated to yield a light yellow oil (11.4 g, 98%).
  • the oil was purified by silica chromatography (225 g Si) eluting with 15% ethyl acetate in heptane followed by 30% ethyl acetate in heptane to yield the product as an oil (9.6 g, 82%).
  • stage 2 (4.2 g, 14 mmol) and TBME (42 mL) and stirred at 40 °C.
  • a solution of (iS)(+)Camphor-io-sulfonic acid (3.2 g, 14 mmol) in warm ethanol (14 mL) was then added in one portion and the solution stirred at RT for 3 hrs.
  • the camphor-sulfonic acid salt was then collected by filtration, washing with TBME (50 mL), and oven dried under vacuum at 40 °C (3.4 g, 91% recovery, 99.4% ee). Freebasing this salt by partition with lM NaOH (100 mL) and TBME (100 mL) yielded the (+) enantiomer of stage 2.
  • the salt (3.6 g) was then recrystallised from a mixture of hot TBME (36 mL, 10 vol) and ethanol (12 mL, 3.3 vol) and oven dried under vacuum at 40 °C (2.8 g, 60% recovery, 97.9% ee). Freebasing this salt by partition with lM NaOH (80 mL) and TBME (80 mL) yielded the (-) enantiomer of stage 2.
  • stage 2 stage 2
  • 2-hydroxypyridine 0.7 g, 7.6 mmol
  • substituted phenethylamine 11.4 mmol
  • the mixture was heated at 165 °C for 4 hrs and cooled to RT.
  • Water (40 mL) and diethyl ether (12 mL) were added and the mixture slurried for 30 minutes.
  • the precipitate was collected by filtration and washed with diethyl ether (20 mL) before being oven dried under vacuum at 45 °C to yield a white solid (2.5 g, 76%).
  • Step 4 Cyclisation A flask was charged with stage 4 (2.5 g, 5.6 mmol) plus toluene (45 mL). POCI3 (1.7 g, 11.3 mmol) was added and the mixture heated to 80 °C for 2 hrs. A gum formed on the flask walls that was subsequently taken into solution by the addition of acetonitrile (10 mL). The solution was heated to 80 °C for a further 2 hours and cooled to 50 °C before the addition of methanol (20 mL). The solvents were removed in vacuo and the residue partitioned between lM NaOH (50 mL) and DCM (50 mL). The organics were dried over Mg-.S04 and evaporated to dryness to give a yellow oil (3 g, assume 100% yield). The crude product plus trimethyl phosphate was used without purification in the following step.
  • the product diastereomers were purified by column chromatography on silica (120 g) eluting with 2% MeOH in DCM then 2% 7N methanolic ammonia in DCM. Clean fractions of the desired stereoisomer (top spot) were combined and evaporated in vacuo to yield an off-white solid (250 mg, 11% yield). Mixed fractions were combined and evaporated in vacuo to give 800 mg of a diastereomer mix enriched in the lower spot (35% yield, -0.5:1 mixture). See experiment tables for approximate purities and stereochemical assignment based on literature precedent (Chem. Comraun., 2014, 50, 1238).
  • a sealed tube was loaded with stage 5 (0.1 mmol) and DMAP (0.4 mmol) in DCM (1 mL).
  • the appropriate alkylating agent was charged (2 eq) and the tube purged with nitrogen, sealed and stirred overnight at room temperature.
  • the mixture was blown to dryness and partitioned between diethyl ether (1 mL) and lM NaOH (1 mL).
  • the organic phase was blown to dryness and columned on a 2g silica cartridge eluting 8 x 5 mL fractions of 1% MeOH / DCM.
  • Product fractions were combined and evaporated to dryness in vacuo to yield an off white solid (40-60% yield).
  • FIG. 11 there is shown the compound represented by formula I (i.e. "HIF-Inhibi"), and three enantiomers (S, R - "UCL-ONY-001"; R, S - “UCL-ONY- 002”; and S, S - “UCL-ONY-003"), and a racemic analogue ("UCL-ONY-004")-
  • HIF-Inhibi three enantiomers
  • S, R - "UCL-ONY-001" R, S - "UCL-ONY- 002"
  • S, S - “UCL-ONY-003 a racemic analogue
  • Figure 12A is a graph showing the percentage inhibition of luciferase activity in U2OS-HRE cells treated with the compounds shown in Figure 11. The compounds were dosed at ⁇ and incubated in 1% 02 for 16 hours.
  • Figure 12B shows Western blot analysis of U2OS-HRE cells treated with compounds as in Figure lC
  • the S, R enantiomer (“UCL-ONY-001”) exhibits similar inhibitory activity as HIF-Inhibi in the U20S_HRE luciferase cells, while UCL-ONY-002, 003 and 004 are inactive.
  • the S, R enantiomer is: (S)-2-(((R)-6,7-dimethoxy- 1,2,3,4- tetrahydroisoquinolin-i-yl)methyl)-3-ethyl-i,6,7,iib-tetrahydro-4H-pyrido[2,i- a]isoquinoline.
  • the S, R enantiomer (“UCL-ONY-001") is believed to be responsible for the activity. This is further confirmed by the mechanism of action analysis shown in Figure 12B, where the inventors have found that the S,R enantiomer (“UCL-ONY-ooi”) has similar inhibitory activity to HIF-Inhibi in blocking HIFia protein induction and eIF-2a phosphorylation.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Epidemiology (AREA)
  • Oncology (AREA)
  • Virology (AREA)
  • Communicable Diseases (AREA)
  • Molecular Biology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Urology & Nephrology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)

Abstract

The invention provides inhibitors of hypoxia-inducible factors (HIF), and their use in the prevention or inhibition of diseases characterised by abnormal HIF activity or levels, such as tumour progression, and the treatment of cancer. The invention encompasses pharmaceutical compositions with a mechanism of action for blocking elevated HIF activity in diseases, such as cancer.

Description

HIF INHIBITORS
The invention relates to hypoxia-inducible factors (HIF), and particularly, although not exclusively, to the inhibition of HIF activity. The invention extends to inhibitors of HIF activity, and their use in the prevention or inhibition of diseases characterised by abnormal HIF activity or levels, such as tumour progression, and the treatment of cancer. The invention encompasses pharmaceutical compositions and methods of treating diseases characterised by elevated HIF activity, such as cancer.
The hypoxia-inducible factor (HIF) transcriptional complex is involved in tumour progression by up-regulating key genes involved in metabolic adaptation, glycolysis (glucose transporters, GLUTi and glycolytic enzymes), proliferation (insulin-like growth factors l and 2) and angiogenesis (VEGF, erythropoietin). HIF is a dimeric transcription factor comprising a regulatory a subunit and constitutively expressed β subunit. HIF-a availability is controlled at the level of protein stability and synthesis by changes in oxygen concentration and growth factors, respectively. Over- expression of HIF-a occurs in most human cancers due to changes in micro- environmental stimuli (e.g. hypoxia, growth factors) and genetic abnormalities that lead to loss of tumour suppressor function (e.g. p53, PTEN, VHL) or oncogenic activation (e.g. Ras, Myc, Src). Thus, targeting HIF function in cancer is an attractive strategy for the development of new anti-cancer agents.
The inventors have developed a cell-based reporter screen (known as "U2OS-HRE- luc") that was used to identify novel small molecule inhibitors of HIF activity. Using this assay, they have now found that one of their hit compounds (which is referred to herein as the compound represented by formula I or simply "formula I" or "HIF-
Inhibi") inhibits both HIF activity and HIF-a expression in response to hypoxia and growth factors in several cancer cell lines. As such, they are the first group to have demonstrated a therapeutic use for the lead compound, which can be used in the treatment or prevention of cancer. In addition, the inventors propose that compound of formula (I) also has use in other settings where blockade of HIF is therapeutically beneficial (e.g. in the hepatitis C viral (HCV) infection life cycle and hepatoma cell migration).
Hence, in a first aspect of the invention, there is provided a compound of formula (I):-
Figure imgf000003_0001
, or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof, for use in therapy or as a medicament.
In a second aspect, there is provided a compound of formula (I), or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof, for use in treating, preventing or ameliorating a disease characterised by abnormal levels of hypoxia-inducible factor (HIF) activity, preferably cancer.
In a third aspect, there is provided a method of treating, preventing or ameliorating a disease characterised by abnormal levels of hypoxia-inducible factor (HIF) activity, preferably cancer, the method comprising administering, to a subject in need of such treatment, a therapeutically effective amount of a compound of formula (I), or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof.
Advantageously, the inventors have shown that the compound of formula (I) not only effectively inhibits HIF activity, but also HIF-a expression in response to hypoxia and growth factors in several cancer cell lines. In addition, they have also found that compound (I) inhibits the growth of a panel of tumour cell lines at submicromolar concentrations. Evaluation of compound (I) showed that it has favourable pharmacokinetic properties in vivo and mice could tolerate a maximum dose of up to loomg/kg daily dosing by intraperitoneal (IP) injection. Based on these promising initial studies, the inventors went on to investigate the effects of compound (I) on growth of PC3 prostate carcinoma cells grown orthotopically. Surprisingly, the inventors found that compound (I) significantly blocked tumour growth and the incidence of metastasis at local and distant lymph nodes. In addition, compound (I) also blocked HIF-ια and VEGF expression in the orthotopic PC3 prostate carcinoma model.
Along with significant findings, the inventors also went on to identify a potential mechanism of action for compound (I) in that it affects key components of the translational machinery that control HIF-a protein synthesis. Compound (I) is structurally similar to emetine, a known protein synthesis inhibitor. However, surprisingly and advantageously, the inventors have found that compound (I) is at least 100-fold less toxic than emetine on tumour cells. Interestingly, previous studies have shown that emetine targets the 40S ribosome at the level of the ribosomal protein S14. Since phosphorylation of the eukaryotic initiation factor eIF-2a regulates translation initiation from the 40S ribosome, the inventors next assessed eIF-2a phosphorylation in response to compound (I). Their initial studies have shown that both emetine and compound of formula (I) block eIF-2a
phosphorylation, suggesting that they may have a similar target profile.
Hence, preferably the compound of formula (I), or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof, inhibits the hypoxia-inducible factor (HIF) transcriptional complex, i.e. it is a HIF pathway inhibitor. The inventors observed that the compound's IC50 for inhibiting HIF activity in the U20S-HRE-luc cell-based assay that was used is in the sub- micromolar range, i.e. ~0.5μΜ.
More preferably, the compound of formula (I), or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof, reduces or blocks expression of hypoxia-inducible factor-i alpha (HIF-ια). The inventors found that the compound's potency in blocking HIF-ια protein induction in hypoxia directly correlates with its IC50 for inhibiting HIF activity, i.e. in the range of about 0.25- 0.5UM.
Preferably, the compound of formula (I), or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof, reduces or blocks expression of vascular endothelial growth factor (VEGF). The compound's potency in blocking VEGF induction in hypoxia directly correlates with its IC50 for inhibiting HIF activity, i.e. ~θ.25-ο.5μΜ. Preferably, the compound of formula (I), or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof, reduces or blocks eIF-2a phosphorylation. The compound's potency in blocking eIF-2a phosphorylation directly correlates with its IC50 for inhibiting HIF activity, i.e. ~θ.25-ο.5μΜ.
The inventors believe that compound of formula (I), or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof, can be used to treat any disease resulting from abnormal levels of HIF or HIF activity. In one
embodiment, abnormal HIF levels may be decreased with respect to those in a healthy individual. However, preferably the disease is characterised by elevated HIF activity with respect to a healthy individual. In some embodiments, in such diseases, HIF is constitutively upregulated and HIF-a (HIF-ia or HIF-2a) protein is overexpressed. For example, the hepatitis C viral (HCV) infection life cycle is known to result in elevated HIF activity, and so hepatitis C can be treated using the compound of formula (I), or a functional analogue, pharmaceutically acceptable salt or solvate thereof.
Compound of formula (I), or a functional analogue, or derivative, or
pharmaceutically acceptable salt or solvate thereof, can be used to treat any tumour or cancer-based disease where HIF is constitutively upregulated and HIF-a (HIF-ia or HIF-2a) protein is overexpressed. For example, the cancer may be a solid tumour or solid cancer. Preferably, compound of formula (I), or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof, is used to treat prostate cancer. Hepatoma cell migration may also be treated.
The skilled person will appreciate that although compound of formula (I) has been demonstrated in the Examples as showing surprising efficacy for inhibiting HIF and therefore exhibits utility for treating tumours and cancers, various functional analogues of compound (I) can also be used, as they can also inhibit HIF. A functional analogue can be defined as being any compound which exhibits at least 8o% HIF inhibition compared to compound (I) using the U20S-HRE-luc cell-based assay without affecting cell viability, i.e. the analogue is not toxic. Toxicity can be defined as being more than 20% cell death within 24 hours, and so functional analogues should not cause more than 20% death.
The inventors have investigated several analogues of compound (I), which are shown in Figures 7-12. For example, the chemical structure of compound (I) can be broken down into three subunits as shown by the double lines in the centre of Figure 8. Arrows ι and 3 in Figure 8 indicate that there are up to 6-iiindependent chemical groups in combination with up to three separate cores resulting in a variety of functional analogues. Accordingly, preferred analogues of compound (I) are shown in Figure 8.
Compound (I), for use, in the invention, may be chiral. Hence, the compound (I) may include any diastereomer and enantiomer of the formula represented by (I). Diastereomers or enantiomers of (I) are believed to display potent HIF inhibitory activity, and such activities may be determined by use of appropriate in vitro and in vivo assays, which will be known to the skilled technician. Compounds defined by formula (I) can therefore include analogues as racemates. Alternatively, the compounds of formula (I) can be pairs of diastereoisomers, or individual
enantiomers, including the threo- and eryi ro-pair of diastereoisomers and the individual threo and erythro enantiomers.
Preferably, the compound (I) is the S, R enantiomer, i.e. (S)-2-(((R)-6,7-dimethoxy- i,2,3,4-tetrahydroisoquinolin-i-yl)methyl)-3-ethyl-i,6,7,iib-tetrahydro-4H- pyrido[2,i-a]isoquinoline.
It will also be appreciated that compounds for use in the invention may also include pharmaceutically active salts, e.g. the hydrochloride.
The inventors have realised that the compound of formula (I) is a surprisingly effective HIF pathway inhibitor.
Hence, in a fourth aspect, there is provided a hypoxia-inducible factor (HIF) pathway inhibitor comprising a compound of formula (I), or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof.
In a fifth aspect, there is provided a compound of formula (I), or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof, for use as hypoxia-inducible factor (HIF) pathway inhibitor. It will be appreciated that the compound of formula (I), or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof according to the invention maybe used in a medicament which may be used in a monotherapy (i.e. use of compound (I) alone), for treating, ameliorating, or preventing a disease characterised by abnormal levels of hypoxia-inducible factor (HIF) activity, preferably cancer. Alternatively, the compound of formula (I), or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof according to the invention may be used as an adjunct to, or in combination with, known therapies for treating, ameliorating, or preventing cancer.
The compound of formula (I), or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof according to the invention may be combined in compositions having a number of different forms depending, in particular, on the manner in which the composition is to be used. Thus, for example, the composition maybe in the form of a powder, tablet, capsule, liquid, ointment, cream, gel, hydrogel, aerosol, spray, micellar solution, transdermal patch, liposome suspension or any other suitable form that may be administered to a person or animal in need of treatment. It will be appreciated that the vehicle of medicaments according to the invention should be one which is well -tolerated by the subject to whom it is given.
Medicaments comprising the compound of formula (I), or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof according to the invention may be used in a number of ways. For instance, oral
administration may be required, in which case the compound may be contained within a composition that may, for example, be ingested orally in the form of a tablet, capsule or liquid. Compositions comprising the compounds of the invention may be administered by inhalation (e.g. intranasally). Compositions may also be formulated for topical use. For instance, creams or ointments may be applied to the skin.
Compounds according to the invention may also be incorporated within a slow- or delayed-release device. Such devices may, for example, be inserted on or under the skin, and the medicament may be released over weeks or even months. The device maybe located at least adjacent the treatment site. Such devices may be particularly advantageous when long-term treatment with compounds used according to the invention is required and which would normally require frequent administration (e.g. at least daily injection). In a preferred embodiment, compounds and compositions according to the invention may be administered to a subject by injection into the blood stream or directly into a site requiring treatment. Injections maybe intravenous (bolus or infusion) or subcutaneous (bolus or infusion), or intradermal (bolus or infusion).
It will be appreciated that the amount of the compound that is required is determined by its biological activity and bioavailability, which in turn depends on the mode of administration, the physiochemical properties of the compound, and whether it is being used as a monotherapy, or in a combined therapy. The frequency of administration will also be influenced by the half-life of the compound within the subject being treated. Optimal dosages to be administered may be determined by those skilled in the art, and will vary with the particular compound in use, the strength of the pharmaceutical composition, the mode of administration, and the advancement of the cancer. Additional factors depending on the particular subject being treated will result in a need to adjust dosages, including subject age, weight, gender, diet, and time of administration.
Generally, in one embodiment, a daily dose of between o.o^g/kg and 500mg/kg of body weight, or between o.img/kg and 200mg/kg body weight of the compound according to the invention may be used for treating, ameliorating, or preventing cancer depending upon which compound or analogue is used.
The compound may be administered before, during or after onset of the cancer to be treated. Daily doses may be given as a single administration (e.g. a single daily injection). Alternatively, the cancer may require administration twice or more times during a day. As an example, compound (I) may be administered as two (or more depending upon the severity of the cancer being treated) daily doses of between 25mg and 7000 mg (i.e. assuming a body weight of 70 kg). A patient receiving treatment may take a first dose upon waking and then a second dose in the evening (if on a two dose regime) or at 3- or 4-hourly intervals thereafter. Alternatively, a slow release device may be used to provide optimal doses of the compounds according to the invention to a patient without the need to administer repeated doses.
Known procedures, such as those conventionally employed by the
pharmaceutical industry (e.g. in vivo experimentation, clinical trials, etc.), may be used to form specific formulations comprising the compounds according to the invention and precise therapeutic regimes (such as daily doses of the compounds and the frequency of administration). The inventors believe that they are the first to describe a pharmaceutical composition for treating cancer, based on the use of the compounds of the invention. Hence, in a sixth aspect of the invention, there is provided a pharmaceutical composition comprising a compound of formula (I), or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof, and a
pharmaceutically acceptable vehicle. The pharmaceutical composition can be used in the therapeutic amelioration, prevention or treatment in a subject of a disease characterised by abnormal levels of hypoxia-inducible factor (HIF) activity, preferably cancer. Thus, the composition is preferably an anti-cancer pharmaceutical composition. Preferably, the compound (I) is (S)-2-(((R)-6,7-dimethoxy-i,2,3,4- tetrahydroisoquinolin-i-yl)methyl)-3-ethyl-i,6,7,iib-tetrahydro-4H-pyrido[2,i- a]isoquinoline.
The invention also provides in a seventh aspect, a process for making the
composition according to the sixth aspect, the process comprising contacting a therapeutically effective amount of a compound of formula (I), or a functional analogue, pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable vehicle. A "subject" may be a vertebrate, mammal, or domestic animal. Hence,
compounds, compositions and medicaments according to the invention maybe used to treat any mammal, for example livestock (e.g. a horse), pets, or may be used in other veterinary applications. Most preferably, however, the subject is a human being.
A "therapeutically effective amount" of compound is any amount which, when administered to a subject, is the amount of drug that is needed to treat the target disease, or produce the desired effect, i.e. inhibits HIF activity.For example, the therapeutically effective amount of compound used maybe from about o.oi mg to about 8oo mg, and preferably from about o.oi mg to about 500 mg. A "pharmaceutically acceptable vehicle" as referred to herein, is any known compound or combination of known compounds that are known to those skilled in the art to be useful in formulating pharmaceutical compositions. In one embodiment, the pharmaceutically acceptable vehicle may be a solid, and the composition may be in the form of a powder or tablet. However, the
pharmaceutical vehicle maybe a liquid, and the pharmaceutical composition is in the form of a solution. Liquid pharmaceutical compositions, which are sterile solutions or suspensions, can be utilized by, for example, intramuscular,
intrathecal, epidural, intraperitoneal, intravenous and particularly subcutaneous injection.
All of the features described herein (including any accompanying claims, abstract and drawings), and/or all of the steps of any method or process so disclosed, maybe combined with any of the above aspects in any combination, except combinations where at least some of such features and/ or steps are mutually exclusive.
For a better understanding of the invention, and to show how embodiments of the same may be carried into effect, reference will now be made, by way of example, to the accompanying Figures, in which:-
Figure l shows that compound I of the invention (referred to herein as "HIF- Inhibi") blocks HIF activity and HIF-a protein induction in hypoxia in a dose- dependent manner without affecting HIF-ιβ or key cellular signaling proteins, ERKi/2 and AKt/PKB. Figure lA: Graph shows HIF (HRE-luciferase) activity measured as relative light units (RLU) in U20S-HRE-luc cells in response to HIF- Inhibi treatment over a dose range as indicated in normoxia or hypoxia for 16 hours. U20S-HRE-luc described in A, were harvested for western blot analysis. Figure lB: Western blots show the effects of HIF-Inhibi on HIF-ια protein in normoxia or hypoxia. Actin was used as a load control. Figure lC: Western blots show HIF-ia, phosphorylated ERK1/2 (ERK1/2-P), and AKT/PKB proteins in the absence (-) and presence of ιμΜ HIF-Inhibi in normoxia (norm) or hypoxia (hyp). Actin was used as a load control. Figure lD: Western blots show the effects of HIF-Inhibi (luM) on HIF-2a protein levels. UT (untreated), and DMSO treated (-) controls are indicated;
Figure 2 shows that HIF-Inhibi blocks the induction of HIF targets (GLUTi and VEGF) and tumour cell migration in hypoxia. Figure 2A: Graph shows vascular endothelial growth factor (VEGF) protein expression measured by ELISA in U2OS- HRE-luc cells in response to HIF-Inhibi treatment over a dose range as indicated in normoxia or hypoxia for 16 hours, Figure 2B: U20S-HRE-luc described in A, were harvested for western blot analysis. Western blots show the effects of HIF-Inhibi on glucose transporteri (GLUTi) protein induction in normoxia or hypoxia. Actin was used as a load control. Figure 2C: Graph shows tumour cell migration (number (no) of migrated cells/field of view) in the absence (-) and presence of 0.5 or 2.5 μΜ HIF- Inibi in normoxia (norm) or hypoxia (hyp) for 16 hours; Figure 3 shows that HIF-Inhibi targets the protein translation machinery. Figure 3A: Western blots show the effects of HIF-Inhibi treatment over a dose range on HIF-ια and phosphoryated eIF-20: (eIF-2a-P) proteins in U20S-HRE-luc cells in normoxia or hypoxia for 16 hours. Actin was used as a load control. Figure 3B:
Western blots show the effects of HIF-Inhibi (ιμΜ) or emetine treatment over a dose range on phosphorylated eIF-20: (eIF-2a-P) in U20S-HRE-luc cells in normoxia or hypoxia for 16 hours. Actin was used as a load control;
Figure 4 shows pharmacodynamic (PD) and pharmacokinetic (PK) effects of 100 mg/kg daily dosing by intraperitoneal (IP) injection of HIF-Inhibi in a human PC3LN5 subcutaneous mouse xenograft model. Figure 4A: Western blots show the effects of control (CT) or HIF-Inhibi treatment (T) on HIF-ια protein levels as a PD endpoint in PC3 tumour xenografts excised from left (L) or right (R) subcutaneous hindlimbs. Actin was used as a load control. Figure 4B: Graph shows levels of HIF- Inhibi (μΜ) in the tumours described in A, measured by LC/MS analyses;
Figure 5 shows that HIF-Inhibi blocks HIF-a, VEGF, tumour growth and metastasis (local and distant) in a human PC3 orthotopic mouse xenograft model. Figure 5A: Western blots shows the effects of control (solv.con) or HIF-Inhibi treatment on the levels of HIF-ια and HIF-ιβ proteins PC3 tumour xenografts excised as indicated at day 16 after 75mg/kg daily dosing by intraperitoneal injection. Figure 5B: Graph shows VEGF protein levels (pg/ml) from pooled tumour xenographs described in A. Figure 5C-F: Graphs show body weight in tumour bearing mice described in A (C), primary tumour weight in grams (g) at day 16 (D), and the weight (g) of local (E) and distant (F) lymph node metastasis. Figure 5G: Graph shows levels of HIF-Inhibi (μΜ) in plasma and pooled tumours described in A, measured by LCMS analyses; Figure 6 shows the chemical structure of HIF-Inhibi according to the invention;
Figure 7 shows the effects of a series of HIF-Inhibi analogues on HIF activity in U20S-HRE-luc cells. Figure 7A: Structures and molecular weights (MW) are shown for a series of chemical analogues (labelled 4-15) of HIF-Inhibi. Figure 7B: Graph shows the effects of DMSO control (1), HIF-Inhibi at ιθμΜ (2), Emetine at θ.θΐδμΜ (3) and analogues (4-15 at ιθμΜ) on HIF activity (relative light units) in the U2OS- HRE-luc cell-based assay in hypoxia (1% 02, for 16 hours);
Figure 8 shows the structures of a panel of functional analogues of HIF-Inhibi that include a variety of different chemical groups as indicated at positions 1, 2 and 3 within the active phamacophore; Figure 9 shows the reaction scheme for synthesising chemical enantiomers and analogues of HIF-Inhibi;
Figure 10 shows the reaction scheme for synthesising 3-dimethylaminomethyl- pentan-2-one methiodide;
Figure 11 shows the purified structures of five of the chemical enantiomers and analogues of HIF-Inhibi which were obtained by using the reaction scheme shown in Figure 9; and Figure 12 shows the effects of the chemical enantiomers and analogues shown in Figure 11 on HIF activity. Figure 12A is a graph showing the percentage inhibition of luciferase activity in U2OS-HRE cells treated with the compounds shown in Figure 11. The compounds were dosed at ΐμΜ and incubated in 1% 02 for 16 hours. Figure 12B shows western blot analysis of U2OS-HRE cells treated with compounds indicated including HIF-Inhibi (HIF-Inh) as in Figure lC to show inhibitory effects on HIF-ια, phosphorylated and total eIF2a protein levels. Tubulin was used as a loading control. All data shown has been either averaged or is representative of 3 independent experiments. Examples
The inventors have found that the compound represented as formula I, which is shown in Figure 6, inhibits both HIF activity and HIF-a expression in response to hypoxia and growth factors in several cancer cell lines. Accordingly, compound (I) can be used therapeutically for the treatment of solid tumours. The compound represented by formula I is known herein as "HIF-Inhibi". Example l - Compound I of the invention blocks HIF-ia protein induction in hypoxia U20S-HRE-luc cells were exposed to normoxia or hypoxia (1% 02) for 16 hours in the presence of DMSO (control) or HIF-Inhibi over a concentration range (0.1-ΐμΜ). Cells were harvested and assessed for HRE-luciferase activity as a measure of HIF activity, and for western blot analysis.
Results
As shown in Figure 1, compound I of the invention blocks HIF activity (HRE- luciferase activity measured as relative light units, RLU) in a dose dependent manner (Figure lA). This dose-dependent inhibitory effect on HIF activity was found to directly correlate with blockade of HIF-a protein induction in hypoxia (Figure lB). The inventors found that compound I of the invention had no significant effect on the expression of key cellular signaling proteins, ERK1/2 and AKt/PKB (Figure iC) at doses which significantly affected HIF, indicating a specific inhibitory effect of compound I of the invention on the HIF pathway. In addition, the inventors show that compound 1 blocks HIF-20 protein induction in hypoxia.
Example 2 - Compound I blocks HIF targets (GLUTi and VEGF) and tumour cell migration in hypoxia
U2OS-HRE-IUC cells were exposed to normoxia or hypoxia (1% 02) for 16 hours in the presence of DMSO (control) or HIF-Inhibi over a concentration range (0.1-ΐμΜ). Cells were harvested and assessed for VEGF and GLUTi protein levels using a quantitative ELISA or by western blot analysis respectively. In addition, tumour cells were exposed to 0.5 or 2.5μΜ HIF-Inhibi in hypoxia, and tumour cell migration was measured using a 2-dimensional filter-based migration assay.
Results
Figure 2 shows that compound I of the invention blocks the induction of HIF target proteins, VEGF and GLUTi (Figure 2A-B) in a dose-dependent manner. These data correlate directly with the dose-dependent inhibitory effects HIF-Inhibi on HIF activity and HIF-ια protein in hypoxia shown in Figure lA-iB. In addition, Figure 2C shows that HIF-Inhibi also reduces tumour cell migration induced in hypoxia in a dose-dependent manner, and is consistent with blockade of the HIF pathway. Example 3 - Compound I targets key components of the protein translation machinery
U20S-HRE-luc cells were exposed to normoxia or hypoxia (1% 02) for 16 hours in the presence of DMSO (control) or HIF-Inhibi over a concentration range (0.1-ΐμΜ). Cells were harvested and components of the protein translational machinery were assessed by western blot analysis.
Results
Figure 3 shows that compound I targets components of the protein translation machinery. The inventors found that HIF-Inhibi blocked eIF-2a phosphorylation in a dose-dependent manner, indicating that compound I affects protein translation. These data correlate directly with the dose-dependent inhibitory effects HIF-Inhibi on HIF activity and HIF-ια protein. Furthermore, the inventors have found that emetine, a known protein translation inhibitor, and analogue of compound 1 also blocks eIF-2a phosphorylation.
Example 4 - Compound I of the invention blocks HIF-ia and shows good
bioavailability in vivo
HIF-Inhibi was administered IP dose of loomg.kg 1 to Nu mice with PC3LN5 xenografts. Mice were killed at 24.I1 and xenografts removed for PD/PK analysis. Tumour samples were homogenised with 3x (v/w) PBS and 5θμΙ, extracted by addition of ΐ5θμΙ, of methanol. Tumour extracts were analysed by LCMS using reverse-phase Synergi Polar-RP (Phenomenx, 50x2.1mm) analytical column and positive ion mode ESI+ MRM.
Results
Figure 4 shows that the concentrations of compound I between left and right flank subcutaneous tumours were comparable. Tumour concentrations ranged between ι.9-35μΜ. Plasma concentrations ranged between 0.07 and 0.3μΜ.
Example 5 - Compound I blocks HIF-q. VEGF. tumour growth and metastasis (local and distant) in a human PCsLNf; orthotopic mouse xenograft model
PC3LN5 (105 cells) were implanted intraprostatically into mice (Nu) and tumours were allowed to develop for 12 days. Mice received HIF-Inhibi (75mg.kg_1) by IP injection daily for 2.5 weeks. Plasma and tumour samples were taken 24h after the last dose and analysed by LCMS. Tumours were excised and homogenised, and assessed for PD endpoints HIF-ια and VEGF proteins. Local and distant lymph node metastases were also evaluated.
Results
Figure 5 shows that compound I blocks HIF-ια and VEGF protein in PC3LN5 orthotopic tumours in vivo. Mouse body weight was not significantly affected over 16 days of daily dosing with HIF-Inhibi, indicating minimal toxicity. HIF-Inhibi significantly blocked tumour growth and metastasis (local and distant) in the PC3LN5 orthotopic xenograft model. HIF-Inhibi showed a good PK profile in tumours, indicating good bioavailability to the tumour.
Example 6 - Chemical structure of compound I of the invention
Referring to Figure 6, there is shown the structure of compound (I), i.e. HIF-Inhibi. Example 7 - Analogues of compound I (batch 1)
A series of analogues (labelled 4-15) of compound I were synthesised, and their structures are shown in Figure 7. U20S-HRE-luc cells were exposed to hypoxia (1% 02) for 16 hours in the presence of DMSO (control), HIF-Inhibi (ιθμΜ), emetine (0.017UM) as positive control, and then each of the analogues (4)-(i5) as shown in Figure 7. Cells were harvested and luciferase activity was measured in cell lysates using a standard luminometer. Data was represented as relative light units (RLU) for each condition.
Results
Figure 7B shows the effects of the compounds on U2OS-HRE luciferase assay, as a measure of HIF activity. HIF-Inhibi and emetine significantly blocked HIF activity in hypoxia, while the analogues tested had minimal inhibitory effects.
Referring to Figure 8, there is shown various other analogues that have been generated, and which show HIF inhibition activity. The chemical structure of compound (I) was broken down into three subunits as shown by the double lines in the centre of the Figure. Arrows 1 and 3 in Figure 8 indicate that there are up to 11 independent chemical groups in combination with up to 3 separate cores (arrow 2) resulting in a variety of functional analogues.
Example 8 - Analogues of compound I (batch 2) A further series of enantiomers and analogues of compound I were synthesised, and their structures are shown in Figure 11. The compounds were synthesised using a six step process, as illustrated in the reaction scheme shown in Figure 9, and explained below. It will be noted that stage 1 of the reaction scheme shown in Figure 9 requires 3-dimethylaminomethyl-pentan-2-one methiodide, which was itself prepared according to the reaction scheme shown in Figure 10.
Synthesis references :-
1. Whittaker N.; Openshaw H. T.; Manufacture of 1, 2, 3, 4, 6, 7-hexahydro-2- oxo-nbh-benzo(a)quinolizines; US 3375254 A.
2. Whittaker N.; The synthesis of emetine and related compounds. Part IX. The use of Wittig-type reagents in the synthesis of 2,3-dehydroemetine; J. Chem. Soc. C, 1969, 94-100.
Brossi A.; Baumann M.; Chopard-dit-Jean L.H.; Wiirsch, J.; Schneider, F.; Schnider O.; Helvetica ChimicaActa, 1959, 42 (3), 772-788.
Synthesis of 3-dimethylaminomethyl-pentan-2-one methiodide
Stage 1 - Condensation
A flask was charged with paraformaldehyde (88 g, 2.9 mol), dimethylamine hydrochloride (150 g, 1.8 mol), pentan-2-one (590 mL, 5.5 mol) and methanol (450 mL). The flask was purged with nitrogen and heated at reflux overnight. The solution was cooled and the pH adjusted to 9 with 2M aqueous NaOH. The product was extracted into diethyl ether (3 x 1400 mL), dried over magnesium sulphate and concentrated in vacuo. The crude mixture was distilled under reduced pressure (vigreux column, 20 torr, head 66 - 74 °C) to obtain -150 mL of a yellow liquid. This was purified by column chromatography on silica (3 kg) eluting with 1% 7N MeOH in DCM and then 2% 7N MeOH in DCM to obtain 58 g of product as a yellow oil (22% yield). Stage 2 - Salt formation
The amine (55 g, 0.4 mol) was filtered under a blanket of nitrogen (to remove oxidation products from storage) into a flask fitted with an overhead stirrer. Ethyl acetate (250 mL) was added and the mixture stirred at RT under nitrogen. Methyl iodide (109 g, 0.8 mol) was then added over 5 minutes with cooling provided to maintain T < 30 °C. The mixture was stirred overnight at RT and then filtered under a blanket of nitrogen washing with ethyl acetate (300 mL). The precipitate was pulled dry on the filter and oven dried under vacuum at 45 °C to obtain 99 g of a white solid (91% yield).
Synthesis of compounds shown in Figure 11
Stage 1 - Cyclisation
A flask was charged with 3-dimethylaminomethyl-pentan-2-one methiodide (58 g, 206 mmol) and dihydroisoquinoline (9 g, 69 mmol) and suspended in ethanol (225 mL). The mixture was heated to reflux under nitrogen overnight. The mixture was cooled to room temperature and filtered. The filter was washed with ethanol (50 mL) and the filtrate combined and concentrated in vacuo to yield a yellow oil (24 g). This was purified by column chromatography on silica (500 g) eluting with 10% ethyl acetate in heptane followed by 20% and 30% ethyl acetate in heptane. The product fractions were combined and the solvent removed in vacuo. The resulting yellow solid was further purified by slurry in ethanol (60 mL) to yield a white solid (7.7 g, 49%). A further crop of product (2.2 g, 14%) was obtained upon concentration of the ethanolic washings to half volume.
Stage 2 - Horner Wadsworth Emmons reaction
A flask was charged with diethyl phthalate (6.9 g, 31 mmol), sodium ethoxide solution (50.3 g, 155 mmol, 21% wt in ethanol), and ethanol (90 mL) and cooled to -5 °C under an atmosphere of nitrogen. Triethyl phosphonoacetate (10.9 g, 49 mmol) was added dropwise maintaining a temperature <5 °C. The solution was allowed to warm to 10 °C and stirred for 1 hr before being cooled to o °C. Stage 1 (8.9 g, 39 mmol) was added in one portion and the mixture stirred for 3 hrs at RT followed by 2 hours at reflux. The ethanol was removed in vacuo and the residue partitioned between toluene (400 mL) and water (400 mL). The phases were separated and the aqueous extracted with a further portion of toluene (50 mL). The combined organics were extracted into lM HC1 (500 mL) which was then basified with NaOH and twice extracted into diethyl ether (2 x 400 mL). The organics were dried over MgS04, filtered and concentrated to yield a light yellow oil (11.4 g, 98%). The oil was purified by silica chromatography (225 g Si) eluting with 15% ethyl acetate in heptane followed by 30% ethyl acetate in heptane to yield the product as an oil (9.6 g, 82%).
Chiral resolution
f +) - camphor-sulfonic acid
A flask was charged with stage 2 (4.2 g, 14 mmol) and TBME (42 mL) and stirred at 40 °C. A solution of (iS)(+)Camphor-io-sulfonic acid (3.2 g, 14 mmol) in warm ethanol (14 mL) was then added in one portion and the solution stirred at RT for 3 hrs. The camphor-sulfonic acid salt was then collected by filtration, washing with TBME (50 mL), and oven dried under vacuum at 40 °C (3.4 g, 91% recovery, 99.4% ee). Freebasing this salt by partition with lM NaOH (100 mL) and TBME (100 mL) yielded the (+) enantiomer of stage 2.
The liquors from the crystallisation were concentrated in vacuo and partitioned between lM NaOH (80 mL) and TBME (80 mL). The organics were dried over MgS04 and concentrated in vacuo to yield the freebase as an oil (2.4 g, 85% (-), 15% (+)). f-) - diparatoluoyl-tartaric acid
A flask was charged with the residue from the first crystallisation (2.4 g, 8 mmol) and TBME (48 mL) and stirred at 40 °C. A solution of diparatoluoyl-L-tartaric acid (3.1 g, 8 mmol) in warm ethanol (8 mL) was then added in one portion and the mixture stirred at RT overnight. The diparatoluoyl-tartaric acid salt was then collected by filtration, washing with TBME (50 mL). The salt (3.6 g) was then recrystallised from a mixture of hot TBME (36 mL, 10 vol) and ethanol (12 mL, 3.3 vol) and oven dried under vacuum at 40 °C (2.8 g, 60% recovery, 97.9% ee). Freebasing this salt by partition with lM NaOH (80 mL) and TBME (80 mL) yielded the (-) enantiomer of stage 2.
The liquors from the crystallisation were concentrated in vacuo and partitioned between lM NaOH (50 mL) and TBME (50 mL). The organics were dried over MgS04 and concentrated in vacuo to yield the freebase as an oil (1.6 g). This was re- subjected to the procedure above to provide an additional 0.2 g of the (+)
enantiomer (99.6% ee) and 0.8 g of the (-) enantiomer (99.3% ee).
Stage 3 - Amide formation
A flask was charged with stage 2 (2.3 g, 7.6 mmol), 2-hydroxypyridine (0.7 g, 7.6 mmol) and the substituted phenethylamine (11.4 mmol). The mixture was heated at 165 °C for 4 hrs and cooled to RT. Water (40 mL) and diethyl ether (12 mL) were added and the mixture slurried for 30 minutes. The precipitate was collected by filtration and washed with diethyl ether (20 mL) before being oven dried under vacuum at 45 °C to yield a white solid (2.5 g, 76%).
Step 4 - Cyclisation A flask was charged with stage 4 (2.5 g, 5.6 mmol) plus toluene (45 mL). POCI3 (1.7 g, 11.3 mmol) was added and the mixture heated to 80 °C for 2 hrs. A gum formed on the flask walls that was subsequently taken into solution by the addition of acetonitrile (10 mL). The solution was heated to 80 °C for a further 2 hours and cooled to 50 °C before the addition of methanol (20 mL). The solvents were removed in vacuo and the residue partitioned between lM NaOH (50 mL) and DCM (50 mL). The organics were dried over Mg-.S04 and evaporated to dryness to give a yellow oil (3 g, assume 100% yield). The crude product plus trimethyl phosphate was used without purification in the following step.
Step , - Hydrogenation
A flask was charged with crude stage 4 (5.6 mmol) in methanol (25 mL). 2M HC1 (25 mL) was added and the flask purged with nitrogen. Platinum (IV) oxide (64 mg, 0.3 mmol) was added and the flask sparged with hydrogen for 6 hrs before being stirred overnight under a head of hydrogen. The mixture was filtered on Celite and the filtrate concentrated in vacuo to remove methanol. The aqueous was basified with 10% Na2C03 and the precipitate collected by filtration (~3 g). The product diastereomers were purified by column chromatography on silica (120 g) eluting with 2% MeOH in DCM then 2% 7N methanolic ammonia in DCM. Clean fractions of the desired stereoisomer (top spot) were combined and evaporated in vacuo to yield an off-white solid (250 mg, 11% yield). Mixed fractions were combined and evaporated in vacuo to give 800 mg of a diastereomer mix enriched in the lower spot (35% yield, -0.5:1 mixture). See experiment tables for approximate purities and stereochemical assignment based on literature precedent (Chem. Comraun., 2014, 50, 1238).
Step 6 - N-alkylation
A sealed tube was loaded with stage 5 (0.1 mmol) and DMAP (0.4 mmol) in DCM (1 mL). The appropriate alkylating agent was charged (2 eq) and the tube purged with nitrogen, sealed and stirred overnight at room temperature. The mixture was blown to dryness and partitioned between diethyl ether (1 mL) and lM NaOH (1 mL). The organic phase was blown to dryness and columned on a 2g silica cartridge eluting 8 x 5 mL fractions of 1% MeOH / DCM. Product fractions were combined and evaporated to dryness in vacuo to yield an off white solid (40-60% yield).
The compounds were analysed and the proton NMR assignments were made as set out in Table 1. Table l: The structure, stereochemistry, LCMS purity and NMR assignments for enantiomers and analogues of compound I
Figure imgf000020_0001
Figure imgf000021_0001
Structure Identifier Stereochemistry LCMS NMR
Purity
0.5Heq, J = 10.8 Hz, 2.7 Hz), 3.53 (dd, o.5Heq, J = 10.8 Hz, 2.7 Hz), 3.65 - 3-73 (m, iH), 3-77 (s, 3H), 3-75 - 3.87 (m, lH), 4.88 - 4.98 (m, lH), 6.61 - 6.66 (m, lH), 6.72 - 6.78 (m, lH), 6.97 - 7-25 (m, 5H)
Example Q - HIF inhibition using Analogues of compound I (batch 2)
Referring to Figure 11, there is shown the compound represented by formula I (i.e. "HIF-Inhibi"), and three enantiomers (S, R - "UCL-ONY-001"; R, S - "UCL-ONY- 002"; and S, S - "UCL-ONY-003"), and a racemic analogue ("UCL-ONY-004")- These compounds were then tested for their HIF activity using the luciferase assay, and the data are shown in Figure 12. Figure 12A is a graph showing the percentage inhibition of luciferase activity in U2OS-HRE cells treated with the compounds shown in Figure 11. The compounds were dosed at ΐμΜ and incubated in 1% 02 for 16 hours. Figure 12B shows Western blot analysis of U2OS-HRE cells treated with compounds as in Figure lC to show inhibitory effects on HIF-ια, phosphorylated and total eIF2a protein levels. Tubulin was used as a loading control.
As can be seen, the S, R enantiomer ("UCL-ONY-001") exhibits similar inhibitory activity as HIF-Inhibi in the U20S_HRE luciferase cells, while UCL-ONY-002, 003 and 004 are inactive. The S, R enantiomer is: (S)-2-(((R)-6,7-dimethoxy- 1,2,3,4- tetrahydroisoquinolin-i-yl)methyl)-3-ethyl-i,6,7,iib-tetrahydro-4H-pyrido[2,i- a]isoquinoline. Therefore, the S, R enantiomer ("UCL-ONY-001") is believed to be responsible for the activity. This is further confirmed by the mechanism of action analysis shown in Figure 12B, where the inventors have found that the S,R enantiomer ("UCL-ONY-ooi") has similar inhibitory activity to HIF-Inhibi in blocking HIFia protein induction and eIF-2a phosphorylation.

Claims

Claims
l. A compound
Figure imgf000024_0001
(I)
, or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof, for use in therapy, or as a medicament.
2. A compound according to claim l, wherein the compound (I) is the S, R enantiomer.
3. A compound according to either claim 1 or claim 2, wherein the compound, or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof, inhibits the hypoxia-inducible factor (HIF) transcriptional complex.
4. A compound according to any preceding claim, wherein the compound, or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof, reduces or blocks expression of hypoxia-inducible factor-i alpha (HIF-ia).
5. A compound according to any preceding claim, wherein the compound, or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof, reduces or blocks expression of vascular endothelial growth factor (VEGF).
6. A compound according to any preceding claim, wherein the compound, or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof, reduces or blocks eIF-2a phosphorylation.
7. A compound of formula (I), or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof according to any one of claims 1- 6, for use in treating, preventing or ameliorating a disease characterised by abnormal levels of hypoxia-inducible factor (HIF) activity, preferably cancer.
8. A compound according to claim 7, wherein the compound, or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof, is used to treat hepatitis C or hepatoma cell migration.
9. A compound according to either claim 7 or 8, wherein the compound, or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof, is used to treat a tumour or cancer-based disease where HIF is constitutively upregulated and HIF-a (HIF-ia or HIF-2a) protein is overexpressed.
10. A compound according to any one of claims 7-9, wherein the cancer is prostate cancer.
11. A hypoxia-inducible factor (HIF) pathway inhibitor comprising a compound of formula (I), or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof according to any one of claims 1-6.
12. A compound of formula (I), or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof according to any one of claims 1- 6, for use as a hypoxia-inducible factor (HIF) pathway inhibitor.
13. A pharmaceutical composition comprising a compound of formula (I), or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof according to any one of claims 1-6, and a pharmaceutically acceptable vehicle.
14. A composition according to claim 13, wherein the composition is an anti- cancer pharmaceutical composition.
15. A process for making the composition according to either claim 13 or 14, the process comprising contacting a therapeutically effective amount of a compound of formula (I), or a functional analogue, or derivative, or pharmaceutically acceptable salt or solvate thereof according to any one of claims 1-6, and a pharmaceutically acceptable vehicle.
PCT/GB2014/053475 2013-11-26 2014-11-25 Hif inhibitors WO2015079213A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US15/039,358 US20170157112A1 (en) 2013-11-26 2014-11-25 Hif inhibitors
EP14803226.1A EP3074013A1 (en) 2013-11-26 2014-11-25 Hif inhibitors
JP2016534721A JP2016538304A (en) 2013-11-26 2014-11-25 HIF inhibitor

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB1320835.0A GB201320835D0 (en) 2013-11-26 2013-11-26 HIF inhibitors
GB1320835.0 2013-11-26

Publications (1)

Publication Number Publication Date
WO2015079213A1 true WO2015079213A1 (en) 2015-06-04

Family

ID=49918212

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2014/053475 WO2015079213A1 (en) 2013-11-26 2014-11-25 Hif inhibitors

Country Status (5)

Country Link
US (1) US20170157112A1 (en)
EP (1) EP3074013A1 (en)
JP (1) JP2016538304A (en)
GB (1) GB201320835D0 (en)
WO (1) WO2015079213A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB993156A (en) * 1961-03-27 1965-05-26 Koninklijke Pharma Fab Nv Quinolizine derivatives
WO2007041294A2 (en) * 2005-09-29 2007-04-12 The Trustees Of Boston University Methods for sensitizing cancer cells to inhibitors
WO2009108360A2 (en) * 2008-02-29 2009-09-03 The Board Of Trustees Of The Leland Stanford Junior University Inhibition of post-radiation tumor growth

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB993156A (en) * 1961-03-27 1965-05-26 Koninklijke Pharma Fab Nv Quinolizine derivatives
WO2007041294A2 (en) * 2005-09-29 2007-04-12 The Trustees Of Boston University Methods for sensitizing cancer cells to inhibitors
WO2009108360A2 (en) * 2008-02-29 2009-09-03 The Board Of Trustees Of The Leland Stanford Junior University Inhibition of post-radiation tumor growth

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
GUPTA RADHEY S ET AL: "Structural Determinants Responsible for the Biological Activity of (-)-Emetine, (-)-Cryptopleurine, and (-)-Tylocrebrine: Structure-Activity Relationship among Related Compounds", MOLECULAR PHARMACOLOGY, AMERICAN SOCIETY FOR PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS, US, vol. 18, no. 1, 1 January 1980 (1980-01-01), pages 136 - 143, XP009181760, ISSN: 0026-895X *
JENNIE A. HICKIN ET AL: "The synthesis and structure revision of NSC-134754", CHEMICAL COMMUNICATIONS, vol. 50, no. 10, 1 January 2014 (2014-01-01), pages 1238, XP055158881, ISSN: 1359-7345, DOI: 10.1039/c3cc48189a *
L C J BAKER ET AL: "The HIF-pathway inhibitor NSC-134754 induces metabolic changes and anti-tumour activity while maintaining vascular function", BRITISH JOURNAL OF CANCER, vol. 106, no. 10, 12 April 2012 (2012-04-12), pages 1638 - 1647, XP055158949, ISSN: 0007-0920, DOI: 10.1038/bjc.2012.131 *
NOAN-MINH CHAU ET AL: "Identification of Novel Small Molecule Inhibitors of Hypoxia- Inducible Factor-1 That Differentially Block Hypoxia-Inducible Factor-1 Activity and Hypoxia-Inducible Factor-1A Induction in Response to Hypoxic Stress and Growth Factors Cell Growth Regulation and Angiogenesis Team, 2 Analytical Technol", CANCER RES, no. 11, 1 June 2005 (2005-06-01), pages 4918 - 4928, XP055158867 *
PAUL KLAUSMEYER ET AL: "Separation and SAR Study of HIF-1[alpha] Inhibitory Tubulosines from Alangium cf. longiflorum", PLANTA MEDICA, vol. 74, no. 3, 1 February 2008 (2008-02-01), pages 258 - 263, XP055158953, ISSN: 0032-0943, DOI: 10.1055/s-2008-1034315 *
WOUTERS ET AL: "Control of the hypoxic response through regulation of mRNA translation", SEMINARS IN CELL AND DEVELOPMENTAL BIOLOGY, ACADEMIC PRESS, GB, vol. 16, no. 4-5, 1 August 2005 (2005-08-01), pages 487 - 501, XP005015255, ISSN: 1084-9521 *
YU-DONG ZHOU ET AL: "Terpenoid Tetrahydroisoquinoline Alkaloids Emetine, Klugine, and Isocephaeline Inhibit the Activation of Hypoxia-Inducible Factor-1 in Breast Tumor Cells", JOURNAL OF NATURAL PRODUCTS, vol. 68, no. 6, 1 June 2005 (2005-06-01), pages 947 - 950, XP055158954, ISSN: 0163-3864, DOI: 10.1021/np050029m *

Also Published As

Publication number Publication date
EP3074013A1 (en) 2016-10-05
JP2016538304A (en) 2016-12-08
GB201320835D0 (en) 2014-01-08
US20170157112A1 (en) 2017-06-08

Similar Documents

Publication Publication Date Title
US9573888B2 (en) Therapeutic compounds
EA023804B1 (en) Oxabicycloheptanes, their preparation and use
US20130281396A1 (en) Treatment of diseases by epigenetic regulation
JP6622205B2 (en) Thienopiperidine derivatives and uses thereof
US9896475B2 (en) Pyridyl analogs of 1-(2-cyano-3,12-dioxooleana-1,9(11)dien-28-oyl) imidazole
TWI597062B (en) Nitrile derivatives and their pharmaceutical use and compositions
AU2013209952A1 (en) New forms and salts of a dihydropyrrolo(1,2-c)imidazolyl aldosterone synthase or aromatase inhibitor
CN111662294A (en) Compound with activity of degrading Btk
US9738613B2 (en) Substituted 1,2,3-triazoles as antitumor agents
EP3043790B1 (en) Novel anthranilic amides and the use thereof
EP3252039B1 (en) Compound containing indoleacetic acid core structure and use thereof
AU2019205595B2 (en) Substituted halo-quinoline derivatives, method of preparation and applications thereof
US9399644B2 (en) [1,3] dioxolo [4,5-G] quinoline-6(5H)thione derivatives as inhibitors of the late SV40 factor (LSF) for use in treating cancer
EP3074013A1 (en) Hif inhibitors
WO2016081589A2 (en) 13-cis-ramba retinamides that degrade mnks for treating cancer
CN106661060A (en) Phenanthroline phosphonic acid derivative and preparation method therefor and application thereof
CN111138449B (en) Preparation of dual-targeting ERK1 and ERK5 inhibitors and anti-tumor application thereof
EP3049400B1 (en) New pi3k/akt/mtor inhibitors and pharmaceutical uses thereof
WO2014066840A1 (en) Aurora kinase inhibitors
EP3013833B1 (en) Tryptophanol-derived oxazoloisoindolinones: small-molecule p53 activators
CN113549046B (en) Bisbecklonin S derivative and preparation method and application thereof
EP3676255A1 (en) Therapeutic indazoles
AU2011274194A1 (en) Phenyl nitrone compounds containing stilbene segment and use thereof
US10058618B2 (en) PAK1-blocking 1,2,3-triazolyl esters
CN117624161A (en) Pyridine carboxylic acid amine derivative and application thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 14803226

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 15039358

Country of ref document: US

ENP Entry into the national phase

Ref document number: 2016534721

Country of ref document: JP

Kind code of ref document: A

REEP Request for entry into the european phase

Ref document number: 2014803226

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2014803226

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: DE